Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 20(8): e1012368, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39172744

RESUMEN

The severity of COVID-19 is linked to excessive inflammation. Neutrophils represent a critical arm of the innate immune response and are major mediators of inflammation, but their role in COVID-19 pathophysiology remains poorly understood. We conducted transcriptomic profiling of neutrophils obtained from patients with mild and severe COVID-19, as well as from SARS-CoV-2 infected mice, in comparison to non-infected healthy controls. In addition, we investigated the inflammasome formation potential in neutrophils from patients and mice upon SARS-CoV-2 infection. Transcriptomic analysis of polymorphonuclear cells (PMNs), consisting mainly of mature neutrophils, revealed a striking type I interferon (IFN-I) gene signature in severe COVID-19 patients, contrasting with mild COVID-19 and healthy controls. Notably, low-density granulocytes (LDGs) from severe COVID-19 patients exhibited an immature neutrophil phenotype and lacked this IFN-I signature. Moreover, PMNs from severe COVID-19 patients showed heightened nigericin-induced caspase1 activation, but reduced responsiveness to exogenous inflammasome priming. Furthermore, IFN-I emerged as a priming stimulus for neutrophil inflammasomes. These findings suggest a potential role for neutrophil inflammasomes in driving inflammation during severe COVID-19. Altogether, these findings open promising avenues for targeted therapeutic interventions to mitigate the pathological processes associated with the disease.


Asunto(s)
COVID-19 , Inflamasomas , Interferón Tipo I , Neutrófilos , SARS-CoV-2 , COVID-19/inmunología , Humanos , Neutrófilos/inmunología , Neutrófilos/metabolismo , Interferón Tipo I/metabolismo , Interferón Tipo I/inmunología , Inflamasomas/inmunología , Inflamasomas/metabolismo , Animales , SARS-CoV-2/inmunología , Ratones , Masculino , Femenino , Persona de Mediana Edad , Inmunidad Innata , Adulto , Ratones Endogámicos C57BL
2.
PLoS Pathog ; 20(7): e1012390, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39038044

RESUMEN

Hantaviruses cause the acute zoonotic diseases hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS). Infected patients show strong systemic inflammation and immune cell activation. NK cells are highly activated in HFRS, suggesting that also other innate lymphoid cells (ILCs) might be responding to infection. Here, we characterized peripheral ILC responses, and measured plasma levels of soluble factors and plasma viral load, in 17 Puumala virus (PUUV)-infected HFRS patients. This revealed an increased frequency of ILC2 in patients, in particular the ILC2 lineage-committed c-Kitlo ILC2 subset. Patients' ILCs showed an activated profile with increased proliferation and displayed altered expression of several homing markers. How ILCs are activated during viral infection is largely unknown. When analyzing PUUV-mediated activation of ILCs in vitro we observed that this was dependent on type I interferons, suggesting a role for type I interferons-produced in response to virus infection-in the activation of ILCs. Further, stimulation of naïve ILC2s with IFN-ß affected ILC2 cytokine responses in vitro, causing decreased IL-5 and IL-13, and increased IL-10, CXCL10, and GM-CSF secretion. These results show that ILCs are activated in HFRS patients and suggest that the classical antiviral type I IFNs are involved in shaping ILC functions.


Asunto(s)
Fiebre Hemorrágica con Síndrome Renal , Inmunidad Innata , Interferón Tipo I , Linfocitos , Fiebre Hemorrágica con Síndrome Renal/inmunología , Fiebre Hemorrágica con Síndrome Renal/virología , Humanos , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Inmunidad Innata/inmunología , Linfocitos/inmunología , Linfocitos/metabolismo , Virus Puumala/inmunología , Masculino , Orthohantavirus/inmunología , Femenino , Adulto , Persona de Mediana Edad , Citocinas/metabolismo , Citocinas/inmunología
3.
Nat Chem Biol ; 19(9): 1054-1062, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37169961

RESUMEN

Preventing the biogenesis of disease-relevant proteins is an attractive therapeutic strategy, but attempts to target essential protein biogenesis factors have been hampered by excessive toxicity. Here we describe KZR-8445, a cyclic depsipeptide that targets the Sec61 translocon and selectively disrupts secretory and membrane protein biogenesis in a signal peptide-dependent manner. KZR-8445 potently inhibits the secretion of pro-inflammatory cytokines in primary immune cells and is highly efficacious in a mouse model of rheumatoid arthritis. A cryogenic electron microscopy structure reveals that KZR-8445 occupies the fully opened Se61 lateral gate and blocks access to the lumenal plug domain. KZR-8445 binding stabilizes the lateral gate helices in a manner that traps select signal peptides in the Sec61 channel and prevents their movement into the lipid bilayer. Our results establish a framework for the structure-guided discovery of novel therapeutics that selectively modulate Sec61-mediated protein biogenesis.


Asunto(s)
Proteínas de la Membrana , Señales de Clasificación de Proteína , Animales , Ratones , Transporte de Proteínas , Proteínas de la Membrana/metabolismo , Canales de Translocación SEC/química , Canales de Translocación SEC/genética , Canales de Translocación SEC/metabolismo , Biosíntesis de Proteínas
5.
PLoS Pathog ; 17(8): e1009843, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34379707

RESUMEN

In humans, orthohantaviruses can cause hemorrhagic fever with renal syndrome (HFRS) or hantavirus pulmonary syndrome (HPS). An earlier study reported that acute Andes virus HPS caused a massive and transient elevation in the number of circulating plasmablasts with specificity towards both viral and host antigens suggestive of polyclonal B cell activation. Immunoglobulins (Igs), produced by different B cell populations, comprise heavy and light chains; however, a certain amount of free light chains (FLCs) is constantly present in serum. Upregulation of FLCs, especially clonal species, associates with renal pathogenesis by fibril or deposit formations affecting the glomeruli, induction of epithelial cell disorders, or cast formation in the tubular network. We report that acute orthohantavirus infection increases the level of Ig FLCs in serum of both HFRS and HPS patients, and that the increase correlates with the severity of acute kidney injury in HFRS. The fact that the kappa to lambda FLC ratio in the sera of HFRS and HPS patients remained within the normal range suggests polyclonal B cell activation rather than proliferation of a single B cell clone. HFRS patients demonstrated increased urinary excretion of FLCs, and we found plasma cell infiltration in archival patient kidney biopsies that we speculate to contribute to the observed FLC excreta. Analysis of hospitalized HFRS patients' peripheral blood mononuclear cells showed elevated plasmablast levels, a fraction of which stained positive for Puumala virus antigen. Furthermore, B cells isolated from healthy donors were susceptible to Puumala virus in vitro, and the virus infection induced increased production of Igs and FLCs. The findings propose that hantaviruses directly activate B cells, and that the ensuing intense production of polyclonal Igs and FLCs may contribute to acute hantavirus infection-associated pathological findings.


Asunto(s)
Lesión Renal Aguda/patología , Linfocitos B/inmunología , Infecciones por Hantavirus/inmunología , Cadenas Ligeras de Inmunoglobulina/sangre , Activación de Linfocitos/inmunología , Orthohantavirus/inmunología , Lesión Renal Aguda/sangre , Lesión Renal Aguda/etiología , Infecciones por Hantavirus/sangre , Infecciones por Hantavirus/virología , Humanos , Cadenas Ligeras de Inmunoglobulina/inmunología
6.
PLoS Pathog ; 17(3): e1009400, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33690725

RESUMEN

Innate immune cells like monocytes patrol the vasculature and mucosal surfaces, recognize pathogens, rapidly redistribute to affected tissues and cause inflammation by secretion of cytokines. We previously showed that monocytes are reduced in blood but accumulate in the airways of patients with Puumala virus (PUUV) caused hemorrhagic fever with renal syndrome (HFRS). However, the dynamics of monocyte infiltration to the kidneys during HFRS, and its impact on disease severity are currently unknown. Here, we examined longitudinal peripheral blood samples and renal biopsies from HFRS patients and performed in vitro experiments to investigate the fate of monocytes during HFRS. During the early stages of HFRS, circulating CD14-CD16+ nonclassical monocytes (NCMs) that patrol the vasculature were reduced in most patients. Instead, CD14+CD16- classical (CMs) and CD14+CD16+ intermediate monocytes (IMs) were increased in blood, in particular in HFRS patients with more severe disease. Blood monocytes from patients with acute HFRS expressed higher levels of HLA-DR, the endothelial adhesion marker CD62L and the chemokine receptors CCR7 and CCR2, as compared to convalescence, suggesting monocyte activation and migration to peripheral tissues during acute HFRS. Supporting this hypothesis, increased numbers of HLA-DR+, CD14+, CD16+ and CD68+ cells were observed in the renal tissues of acute HFRS patients compared to controls. In vitro, blood CD16+ monocytes upregulated CD62L after direct exposure to PUUV whereas CD16- monocytes upregulated CCR7 after contact with PUUV-infected endothelial cells, suggesting differential mechanisms of activation and response between monocyte subsets. Together, our findings suggest that NCMs are reduced in blood, potentially via CD62L-mediated attachment to endothelial cells and monocytes are recruited to the kidneys during HFRS. Monocyte mobilization, activation and functional impairment together may influence the severity of disease in acute PUUV-HFRS.


Asunto(s)
Fiebre Hemorrágica con Síndrome Renal/sangre , Fiebre Hemorrágica con Síndrome Renal/inmunología , Monocitos/inmunología , Adulto , Anciano , Femenino , Humanos , Riñón/inmunología , Masculino , Persona de Mediana Edad , Virus Puumala
7.
PLoS Pathog ; 17(7): e1009721, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34228753

RESUMEN

Severe COVID-19 is characterized by extensive pulmonary complications, to which host immune responses are believed to play a role. As the major arm of innate immunity, neutrophils are one of the first cells recruited to the site of infection where their excessive activation can contribute to lung pathology. Low-density granulocytes (LDGs) are circulating neutrophils, whose numbers increase in some autoimmune diseases and cancer, but are poorly characterized in acute viral infections. Using flow cytometry, we detected a significant increase of LDGs in the blood of acute COVID-19 patients, compared to healthy controls. Based on their surface marker expression, COVID-19-related LDGs exhibit four different populations, which display distinctive stages of granulocytic development and most likely reflect emergency myelopoiesis. Moreover, COVID-19 LDGs show a link with an elevated recruitment and activation of neutrophils. Functional assays demonstrated the immunosuppressive capacities of these cells, which might contribute to impaired lymphocyte responses during acute disease. Taken together, our data confirms a significant granulocyte activation during COVID-19 and suggests that granulocytes of lower density play a role in disease progression.


Asunto(s)
COVID-19/inmunología , Granulocitos/clasificación , Enfermedad Aguda , Adulto , Anciano , COVID-19/sangre , Estudios de Casos y Controles , Estudios de Cohortes , Convalecencia , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Granulocitos/citología , Humanos , Tolerancia Inmunológica/inmunología , Masculino , Persona de Mediana Edad , Receptores Depuradores de Clase E/análisis , Índice de Severidad de la Enfermedad
8.
Cytometry A ; 103(4): 313-324, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36279192

RESUMEN

Though cryopreservation of cell fractions is widely used in flow cytometry studies, whole blood cryopreservation is more challenging due to the presence of erythrocytes and effects of fixatives commonly used for preservation. Here, we evaluated and compared head-to-head the performance of four commercial whole blood cryopreservation kits; (1) Cytodelics, (2) Stable-Lyse V2 and Stable-Store V2 (SLSS-V2), (3) Proteomic stabilizer (PROT-1), and (4) Transfix. We found that PROT-1, Transfix, and Cytodelics maintained the distribution of major leukocyte subsets-granulocytes, T cells, natural killer cells, and B cells, on a comparable level to unpreserved samples, despite the attenuation of fluorescence intensities in flow cytometric assays. Moreover, these three stabilizers also maintained the activated phenotypes of neutrophils upon stimulation with N-formylmethionyl-leucyl-phenylalanine and lipopolysaccharides. The upregulation of adhesion molecules (CD11b), Fc receptors (CD16), and granule proteins (CD66b), as well as the shedding of surface L-selectin (CD62L), was conserved most efficiently in PROT-1 and Cytodelics when compared to samples only treated with erythrocyte lysing. However, none of the stabilizers provided a reliable detection of CCR7 for accurate quantification of T cell maturation stages. We also evaluated the performance of Cytodelics in longitudinal clinical samples obtained from acute COVID-19 patients, where it allowed reliable detection of lymphopenia and granulocyte expansion. These results support the feasibility of whole blood cryopreservation for immunophenotyping by flow cytometry, particularly in longitudinal studies. In conclusion, the performance of different stabilizers is variable and therefore the choice of stabilizers should depend on cell type of interest, as well as antibody clones and experimental design of each study.


Asunto(s)
COVID-19 , Proteómica , Humanos , Citometría de Flujo , Leucocitos , Granulocitos
9.
Emerg Infect Dis ; 28(6): 1286-1288, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35608951

RESUMEN

We report an experimental infection of American mink with SARS-CoV-2 Omicron variant and show that mink remain positive for viral RNA for days, experience clinical signs and histopathologic changes, and transmit the virus to uninfected recipients. Preparedness is crucial to avoid spread among mink and spillover to human populations.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , COVID-19/veterinaria , Humanos , Visón
10.
Glia ; 69(12): 2947-2962, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34427356

RESUMEN

The function of astrocytes intertwines with the extracellular matrix, whose neuron and glial cell-derived components shape neuronal plasticity. Astrocyte abnormalities have been reported in the brain of the mouse model for fragile X syndrome (FXS), the most common cause of inherited intellectual disability, and a monogenic cause of autism spectrum disorder. We compared human FXS and control astrocytes generated from human induced pluripotent stem cells and we found increased expression of urokinase plasminogen activator (uPA), which modulates degradation of extracellular matrix. Several pathways associated with uPA and its receptor function were activated in FXS astrocytes. Levels of uPA were also increased in conditioned medium collected from FXS hiPSC-derived astrocyte cultures and correlated inversely with intracellular Ca2+ responses to activation of L-type voltage-gated calcium channels in human astrocytes. Increased uPA augmented neuronal phosphorylation of TrkB within the docking site for the phospholipase-Cγ1 (PLCγ1), indicating effects of uPA on neuronal plasticity. Gene expression changes during neuronal differentiation preceding astrogenesis likely contributed to properties of astrocytes with FXS-specific alterations that showed specificity by not affecting differentiation of adenosine triphosphate (ATP)-responsive astrocyte population. To conclude, our studies identified uPA as an important regulator of astrocyte function and demonstrated that increased uPA in human FXS astrocytes modulated astrocytic responses and neuronal plasticity.


Asunto(s)
Trastorno del Espectro Autista , Síndrome del Cromosoma X Frágil , Células Madre Pluripotentes Inducidas , Animales , Astrocitos/metabolismo , Trastorno del Espectro Autista/metabolismo , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
11.
Euro Surveill ; 25(11)2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32209163

RESUMEN

The first case of coronavirus disease (COVID-19) in Finland was confirmed on 29 January 2020. No secondary cases were detected. We describe the clinical picture and laboratory findings 3-23 days since the first symptoms. The SARS-CoV-2/Finland/1/2020 virus strain was isolated, the genome showing a single nucleotide substitution to the reference strain from Wuhan. Neutralising antibody response appeared within 9 days along with specific IgM and IgG response, targeting particularly nucleocapsid and spike proteins.


Asunto(s)
Trazado de Contacto , Infecciones por Coronavirus , Coronavirus/genética , Coronavirus/aislamiento & purificación , Pandemias , Neumonía Viral , Síndrome Respiratorio Agudo Grave/inmunología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología , Viaje , Adulto , Anticuerpos Antivirales/sangre , Infecciones Asintomáticas , Betacoronavirus , COVID-19 , Prueba de COVID-19 , China , Técnicas de Laboratorio Clínico , Coronavirus/inmunología , Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/transmisión , Infecciones por Coronavirus/virología , Femenino , Finlandia , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoglobulina A/sangre , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Pruebas de Neutralización , Neumonía Viral/diagnóstico , Neumonía Viral/transmisión , Neumonía Viral/virología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , SARS-CoV-2 , Síndrome Respiratorio Agudo Grave/etiología , Síndrome Respiratorio Agudo Grave/virología , Proteínas del Envoltorio Viral
12.
J Infect Dis ; 213(10): 1632-41, 2016 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-26704613

RESUMEN

Hantaviruses are zoonotic viruses that show various degrees of vasculopathy in humans. In this study, we analyzed the regulation of 2 fibrinolytic parameters, tissue plasminogen activator (tPA) and its physiological inhibitor, plasminogen activator inhibitor 1 (PAI-1), in Puumala hantavirus (PUUV)-infected patients and in human microvascular endothelial cells. We detected strong upregulation of tPA in the acute phase of illness and in PUUV-infected macaques and found the tPA level to positively correlate with disease severity. The median levels of PAI-1 during the acute stage did not differ from those during the recovery phase. In concordance, hantaviruses induced tPA but not PAI-1 in microvascular endothelial cells, and the induction was demonstrated to be dependent on type I interferon. Importantly, type I and II interferons directly upregulated tPA through signal transducer and activator of transcription 1 (STAT1), which regulated tPA gene expression via a STAT1-responsive enhancer element. These results suggest that tPA may be a general factor in the immunological response to viruses.


Asunto(s)
Fiebre Hemorrágica con Síndrome Renal/virología , Interferón Tipo I/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Virus Puumala/patogenicidad , Factor de Transcripción STAT1/metabolismo , Activador de Tejido Plasminógeno/metabolismo , Animales , Chlorocebus aethiops , Estudios de Cohortes , Células Endoteliales/metabolismo , Regulación Viral de la Expresión Génica , Fiebre Hemorrágica con Síndrome Renal/metabolismo , Humanos , Interferón Tipo I/genética , Macaca fascicularis , Inhibidor 1 de Activador Plasminogénico/genética , Factor de Transcripción STAT1/genética , Transducción de Señal , Activador de Tejido Plasminógeno/genética , Regulación hacia Arriba , Células Vero , Factores de Virulencia
13.
J Gen Virol ; 97(5): 1052-1059, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26916544

RESUMEN

Puumala virus (PUUV) grows slowly in cell culture. To study antigenic properties of PUUV, an amenable method for their expression would be beneficial. To achieve this, a replication-defective recombinant vesicular stomatitis virus, rVSVΔG*EGFP, was rescued using BSRT7/5 and encephalomyocarditis virus (EMCV) internal ribosomal entry site (IRES)-enabled rescue plasmids. Using these particles, pseudotypes bearing PUUV Sotkamo strain glycoproteins were produced, with titres in the range 105-108, and were used in pseudotype focus reduction neutralization tests (pFRNTs) with neutralizing monoclonal antibodies and patient sera. The results were compared with those from orthodox focus reduction neutralization tests (oFRNTs) using native PUUV with the same samples and showed a strong positive correlation (rs = 0.82) between the methods. While developing the system we identified three amino acids which were mutated in the Vero E6 cell culture adapted PUUV prototype Sotkamo strain sequence, and changing these residues was critical for expression and neutralizing antibody binding of PUUV glycoproteins.


Asunto(s)
Glicoproteínas/metabolismo , Pruebas de Neutralización/métodos , Virus Puumala/metabolismo , Virus Vaccinia/fisiología , Virus de la Estomatitis Vesicular Indiana/fisiología , Replicación Viral/fisiología , Glicoproteínas/química , Glicoproteínas/clasificación , Modelos Moleculares , Conformación Proteica , Virus de la Estomatitis Vesicular Indiana/clasificación , Virus de la Estomatitis Vesicular Indiana/genética
14.
Biol Lett ; 12(9)2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27677814

RESUMEN

Trade-offs in the allocation of finite-energy resources among immunological defences and other physiological processes are believed to influence infection risk and disease severity in food-limited wildlife populations. However, this prediction has received little experimental investigation. Here we test the hypothesis that food limitation impairs the ability of wild field voles (Microtus agrestis) to mount an immune response against parasite infections. We conducted a replicated experiment on vole populations maintained in large outdoor enclosures during boreal winter, using food supplementation and anthelmintic treatment of intestinal nematodes. Innate immune responses against intestinal parasite infections were compared between food-supplemented and non-supplemented voles. Voles with high food availability mounted stronger immune responses against intestinal nematode infections than food-limited voles. No food effects were seen in immune responses to intracellular coccidian parasites, possibly owing to their ability to avoid activation of innate immune pathways. Our findings demonstrate that food availability constrains vole immune responses against nematode infections, and support the concept that spatio-temporal heterogeneity in food availability creates variation in infectious disease susceptibility.

15.
J Gen Virol ; 95(Pt 11): 2356-2364, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25013204

RESUMEN

Hantaviruses are zoonotic viruses that cause life-threatening diseases when transmitted to humans. Severe hantavirus infection is manifested by impairment of renal function, pulmonary oedema and capillary leakage. Both innate and adaptive immune responses contribute to the pathogenesis, but the underlying mechanisms are not fully understood. Here, we showed that galectin-3-binding protein (Gal-3BP) was upregulated as a result of hantavirus infection both in vitro and in vivo. Gal-3BP is a secreted glycoprotein found in human serum, and increased Gal-3BP levels have been reported in chronic viral infections and in several types of cancer. Our in vitro experiments showed that, whilst Vero E6 cells (an African green monkey kidney cell line) constitutively expressed and secreted Gal-3BP, this protein was detected in primary human cells only as a result of hantavirus infection. Analysis of Gal-3BP levels in serum samples of cynomolgus macaques infected experimentally with hantavirus indicated that hantavirus infection induced Gal-3BP also in vivo. Finally, analysis of plasma samples collected from patients hospitalized because of acute hantavirus infection showed higher Gal-3BP levels during the acute than the convalescent phase. Furthermore, the Gal-3BP levels in patients with haemorrhagic fever with renal syndrome correlated with increased complement activation and with clinical variables reflecting the severity of acute hantavirus infection.


Asunto(s)
Antígenos de Neoplasias/biosíntesis , Biomarcadores de Tumor/biosíntesis , Proteínas Portadoras/biosíntesis , Galectina 3/metabolismo , Glicoproteínas/biosíntesis , Infecciones por Hantavirus/metabolismo , Enfermedad Aguda , Animales , Antígenos de Neoplasias/sangre , Antígenos Virales/metabolismo , Biomarcadores de Tumor/sangre , Proteínas Portadoras/sangre , Chlorocebus aethiops , Activación de Complemento , Femenino , Glicoproteínas/sangre , Infecciones por Hantavirus/inmunología , Infecciones por Hantavirus/virología , Fiebre Hemorrágica con Síndrome Renal/inmunología , Fiebre Hemorrágica con Síndrome Renal/metabolismo , Fiebre Hemorrágica con Síndrome Renal/virología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Macaca fascicularis , Masculino , Virus Puumala , Distribución Tisular , Células Vero
16.
J Neuroimmunol ; 395: 578432, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39151321

RESUMEN

The coagulation cascade and fibrinolysis have links with neuroinflammation and increased activation of the coagulation system has been reported in MS patients. We quantified levels of D-dimer, tissue plasminogen activator (tPA), plasminogen activator inhibitor-1 (PAI-1) and the bioactivity of bacterial lipopolysaccharide (LPS) in cerebrospinal fluid (CSF) and plasma from newly diagnosed untreated MS patients and controls. These molecules showed multiple correlations with each other as well as with age, HLA-DRB1*15:01, body-mass-index and CSF IgG. Our results confirm previous findings of increased plasma PAI-1 and LPS in MS patients compared to controls indicating changes in platelet function and gut permeability in MS.


Asunto(s)
Fibrinólisis , Lipopolisacáridos , Esclerosis Múltiple , Inhibidor 1 de Activador Plasminogénico , Humanos , Femenino , Masculino , Adulto , Esclerosis Múltiple/líquido cefalorraquídeo , Esclerosis Múltiple/sangre , Inhibidor 1 de Activador Plasminogénico/líquido cefalorraquídeo , Inhibidor 1 de Activador Plasminogénico/sangre , Persona de Mediana Edad , Fibrinólisis/fisiología , Activador de Tejido Plasminógeno/líquido cefalorraquídeo , Activador de Tejido Plasminógeno/sangre , Productos de Degradación de Fibrina-Fibrinógeno/análisis , Productos de Degradación de Fibrina-Fibrinógeno/metabolismo , Adulto Joven
17.
Front Immunol ; 15: 1419787, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39011044

RESUMEN

Puumala orthohantavirus-caused hemorrhagic fever with renal syndrome (PUUV-HFRS) is characterized by strong neutrophil activation. Neutrophils are the most abundant immune cell type in the circulation and are specially equipped to rapidly respond to infections. They are more heterogenous than previously appreciated, with specific neutrophil subsets recently implicated in inflammation and immunosuppression. Furthermore, neutrophils can be divided based on their density to either low-density granulocytes (LDGs) or "normal density" polymorphonuclear cell (PMN) fractions. In the current study we aimed to identify and characterize the different neutrophil subsets in the circulation of PUUV-HFRS patients. PMNs exhibited an activation of antiviral pathways, while circulating LDGs were increased in frequency following acute PUUV-HFRS. Furthermore, cell surface marker expression analysis revealed that PUUV-associated LDGs are primarily immature and most likely reflect an increased neutrophil production from the bone marrow. Interestingly, both the frequency of LDGs and the presence of a "left shift" in blood associated with the extent of thrombocytopenia, one of the hallmarks of severe HFRS, suggesting that maturing neutrophils could play a role in disease pathogenesis. These results imply that elevated circulating LDGs might be a general finding in acute viral infections. However, in contrast to the COVID-19 associated LDGs described previously, the secretome of PUUV LDGs did not show significant immunosuppressive ability, which suggests inherent biological differences in the LDG responses that can be dependent on the causative virus or differing infection kinetics.


Asunto(s)
Fiebre Hemorrágica con Síndrome Renal , Neutrófilos , Virus Puumala , Trombocitopenia , Fiebre Hemorrágica con Síndrome Renal/inmunología , Fiebre Hemorrágica con Síndrome Renal/virología , Neutrófilos/inmunología , Humanos , Trombocitopenia/inmunología , Trombocitopenia/virología , Virus Puumala/inmunología , Masculino , Persona de Mediana Edad , Femenino , Adulto , Activación Neutrófila , Anciano
18.
Microbiol Spectr ; 12(4): e0419922, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38363137

RESUMEN

In response to the ongoing COVID-19 pandemic, the quest for coronavirus inhibitors has inspired research on a variety of small proteins beyond conventional antibodies, including robust single-domain antibody fragments, i.e., "nanobodies." Here, we explore the potential of nanobody engineering in the development of antivirals and diagnostic tools. Through fusion of nanobody domains that target distinct binding sites, we engineered multimodular nanobody constructs that neutralize wild-type SARS-CoV-2 and the Alpha and Delta variants at high potency, with IC50 values as low as 50 pM. Despite simultaneous binding to distinct epitopes, Beta and Omicron variants were more resistant to neutralization by the multimodular nanobodies, which highlights the importance of accounting for antigenic drift in the design of biologics. To further explore the applications of nanobody engineering in outbreak management, we present an assay based on fusions of nanobodies with fragments of NanoLuc luciferase that can detect sub-nanomolar quantities of the SARS-CoV-2 spike protein in a single step. Our work showcases the potential of nanobody engineering to combat emerging infectious diseases. IMPORTANCE: Nanobodies, small protein binders derived from the camelid antibody, are highly potent inhibitors of respiratory viruses that offer several advantages over conventional antibodies as candidates for specific therapies, including high stability and low production costs. In this work, we leverage the unique properties of nanobodies and apply them as building blocks for new therapeutic and diagnostic tools. We report ultra-potent SARS-CoV-2 inhibition by engineered nanobodies comprising multiple modules in structure-guided combinations and develop nanobodies that carry signal molecules, allowing rapid detection of the SARS-CoV-2 spike protein. Our results highlight the potential of engineered nanobodies in the development of effective countermeasures, both therapeutic and diagnostic, to manage outbreaks of emerging viruses.


Asunto(s)
COVID-19 , Anticuerpos de Dominio Único , Glicoproteína de la Espiga del Coronavirus , Humanos , SARS-CoV-2/genética , COVID-19/diagnóstico , Pandemias , Anticuerpos de Dominio Único/genética , Anticuerpos Neutralizantes , Anticuerpos Antivirales
19.
Virus Res ; 341: 199315, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38211733

RESUMEN

Prolonged T cell lymphopenia is common in COVID-19, caused by SARS-CoV-2. While the mechanisms of lymphopenia during COVID-19 remain elusive, it is especially pronounced in a specialized innate-like T cell population called Mucosal Associated Invariant T cells (MAITs). MAITs has been suggested to express Angiotensin-Converting Enzyme 2 (ACE2), which is the well-known cellular receptor for SARS-CoV-2. However, it is still unclear if SARS-CoV-2 can infect or affect MAIT cells directly. In this study, we performed multicolor flow cytometry on peripheral blood mononuclear cells obtained from COVID-19 patients to assess the frequencies of CD8+Vα7.2+CD161+ MAIT subsets at acute and convalescent disease phases. The susceptibility of MAITs and T cells to direct exposure by SARS-CoV-2 was analysed using cells isolated from healthy donor buffy coats by viability assays, virus-specific RT-PCR, and flow cytometry. In situ lung immunofluorescence was used to evaluate retention of T cells, especially MAIT cells, in lung tissues during acute COVID-19. Our study confirms previous reports indicating that circulating MAITs are activated, and their frequency is declined in patients with acute SARS-CoV-2 infection, whereas an accumulation of MAITs and T cells was seen in the lung tissue of individuals with fatal COVID-19. However, despite a fraction of MAITs found to express ACE2, no evidence for the susceptibility of MAITs for direct infection or activation by SARS-CoV-2 particles was observed. Thus, their activation and decline in the circulation is most likely explained by indirect mechanisms involving other immune cells and cytokine-induced pro-inflammatory environment but not by direct exposure to viral particles at the infection site.


Asunto(s)
COVID-19 , Linfopenia , Células T Invariantes Asociadas a Mucosa , Humanos , Enzima Convertidora de Angiotensina 2 , Leucocitos Mononucleares , SARS-CoV-2 , Pulmón
20.
Proc Natl Acad Sci U S A ; 107(6): 2425-30, 2010 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-20145116

RESUMEN

Vascular endothelial growth factors (VEGFs) regulate blood and lymph vessel formation through activation of three receptor tyrosine kinases, VEGFR-1, -2, and -3. The extracellular domain of VEGF receptors consists of seven immunoglobulin homology domains, which, upon ligand binding, promote receptor dimerization. Dimerization initiates transmembrane signaling, which activates the intracellular tyrosine kinase domain of the receptor. VEGF-C stimulates lymphangiogenesis and contributes to pathological angiogenesis via VEGFR-3. However, proteolytically processed VEGF-C also stimulates VEGFR-2, the predominant transducer of signals required for physiological and pathological angiogenesis. Here we present the crystal structure of VEGF-C bound to the VEGFR-2 high-affinity-binding site, which consists of immunoglobulin homology domains D2 and D3. This structure reveals a symmetrical 22 complex, in which left-handed twisted receptor domains wrap around the 2-fold axis of VEGF-C. In the VEGFs, receptor specificity is determined by an N-terminal alpha helix and three peptide loops. Our structure shows that two of these loops in VEGF-C bind to VEGFR-2 subdomains D2 and D3, while one interacts primarily with D3. Additionally, the N-terminal helix of VEGF-C interacts with D2, and the groove separating the two VEGF-C monomers binds to the D2/D3 linker. VEGF-C, unlike VEGF-A, does not bind VEGFR-1. We therefore created VEGFR-1/VEGFR-2 chimeric proteins to further study receptor specificity. This biochemical analysis, together with our structural data, defined VEGFR-2 residues critical for the binding of VEGF-A and VEGF-C. Our results provide significant insights into the structural features that determine the high affinity and specificity of VEGF/VEGFR interactions.


Asunto(s)
Factor A de Crecimiento Endotelial Vascular/química , Factor C de Crecimiento Endotelial Vascular/química , Receptor 2 de Factores de Crecimiento Endotelial Vascular/química , Animales , Sitios de Unión/genética , Línea Celular , Supervivencia Celular , Cristalografía por Rayos X , Humanos , Enlace de Hidrógeno , Cinética , Modelos Moleculares , Mutación , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Spodoptera , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor C de Crecimiento Endotelial Vascular/genética , Factor C de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/química , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/química , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA